You are on page 1of 10

REVIEWS

C I R C A D I A N R H Y T H M S I N E N D O C R I N O L O G Y A N D M E TA B O L I S M

The human stress response


Georgina Russell* and Stafford Lightman   *
Abstract | The human stress response has evolved to maintain homeostasis under conditions of
real or perceived stress. This objective is achieved through autoregulatory neural and hormonal
systems in close association with central and peripheral clocks. The hypothalamic–pituitary–
adrenal axis is a key regulatory pathway in the maintenance of these homeostatic processes.
The end product of this pathway — cortisol — is secreted in a pulsatile pattern, with changes
in pulse amplitude creating a circadian pattern. During acute stress, cortisol levels rise and
pulsatility is maintained. Although the initial rise in cortisol follows a large surge in adrenocortico­
tropic hormone levels, if long-​term inflammatory stress occurs, adrenocorticotropic hormone
levels return to near basal levels while cortisol levels remain raised as a result of increased adrenal
sensitivity. In chronic stress, hypothalamic activation of the pituitary changes from corticotropin-​
releasing hormone-​dominant to arginine vasopressin-​dominant, and cortisol levels remain raised
due at least in part to decreased cortisol metabolism. Acute elevations in cortisol levels are
beneficial to promoting survival of the fittest as part of the fight-​or-flight response. However,
chronic exposure to stress results in reversal of the beneficial effects, with long-​term cortisol
exposure becoming maladaptive, which can lead to a broad range of problems including the
metabolic syndrome, obesity , cancer, mental health disorders, cardiovascular disease and
increased susceptibility to infections. Neuroimmunoendocrine modulation in disease states
and glucocorticoid-​based therapeutics are also discussed.

Zeitgebers
In response to a stressor, the body activates multiple coor­ to optimize tissue-​specific glucocorticoid signalling
Cues that entrain or dinated and dynamic processes to restore homeo­stasis, and maintain the rapid reactivity necessary for a stress
synchronize the body’s preserve life and ultimately achieve evolutionary success response system.
24-h cycle for the species. The importance of endocrine systems
in this homeostatic regulation has been known since Circadian clocks
Ultradian rhythms
Biological rhythms that occur the early studies of Hans Selye1 in the 1930s, when acti­ In the absence of internal or external stressors, the
with a frequency of <24 h. vation of the sympatho–adrenomedullary (SAM) and integrity of physiological systems is maintained in a
hypothalamic–pituitary–adrenal (HPA) axes was des­ dynamic fashion over 24 h by an internal circadian clock
Circadian clock cribed in response to physical injury and exertion as well that antici­pates the changes occurring over the 24-h day.
A biochemical oscillator
with phases synchronized with
as perceived psychological threats. Interestingly, antici­ In the past, the neurocentric hierarchical view was that
solar time. pation of these threats is itself a very potent activator of body rhythms were controlled from a master clock in the
these systems2. Homeostatic processes also interact with hypothalamic suprachiasmatic nucleus (SCN)6. Now, it
internal and external Zeitgebers such as the light–dark is very clear that peripheral clocks also exist in most, if
cycle and internal body clocks3,4. These internal clocks not all, tissues of the body, which have their own auton­
enable the body to anticipate regular changes in the omous transcriptional autoregulatory feedback loops7.
environment to ensure optimal fitness across the 24 h The core clock genes, both in the SCN and peripheral
and thus the best chance for survival5. The human stress clocks, are CLOCK and BMAL1. Activation of these
Translational Health Sciences, response is an additional homeostatic mechanism that genes generates a heterodimer of BMAL1 with either
Dorothy Hodgkin Building, provides a better chance of survival when the body is CLOCK or NPAS2, which bind at promoter elements
Bristol Medical School, under threat and mobilizes neural and hormonal net­ called E-​boxes to drive genes encoding period 1–3, crypto­
University of Bristol, works to optimize cognitive, cardiovascular, immuno­ chrome 1–2 and nuclear receptor subfamily 1–2. The
Bristol, UK.
logical and metabolic function (Fig. 1). In this Review we resultant proteins then feedback to repress BMAL1 by a
*e-​mail: Georgina.russell@
discuss how the HPA axis helps achieve and maintain series of feedback loops, generating a 24-h rhythm8,9 that
bristol.ac.uk; Stafford.
Lightman@bristol.ac.uk homeostasis. We will show how it utilizes rhythmic 24-h allows time-​of-day-​dependent regulation of downstream
https://doi.org/10.1038/ patterns of secretion to achieve appropriate tissue activity genomic pathways. This rhythmicity is crucial for homeo­
s41574-019-0228-0 at different times of the day, and faster ultradian rhythms stasis. The body can only behave optimally when all

Nature Reviews | Endocrinology


Reviews

Key points
inhibit corticotropin-​releasing hormone (CRH) and
arginine vasopressin (AVP) release during the inactive
• The hypothalamic–pituitary–adrenal (HPA) axis is a key system that synchronizes the phase of the cycle31,32. Furthermore, to produce appro­
stress response with circadian regulatory processes. priate reactivity to physiological, cognitive and emo­
• Regulation of the HPA axis is very dynamic with both ultradian and circadian tional stressors, the brain stem and limbic system also
oscillations. modulate HPA activity via projections to the PVN33,34.
• Short-​term and longer-​term stress result in different regulatory mechanisms involving Axonal terminals in the median eminence release
hypothalamic, pituitary and adrenal activity, as well as cortisol metabolism. CRH and AVP into the hypophyseal portal system where
• Chronic elevation and nonphysiological patterns of cortisol result in poor cognitive, they are transported to the pituitary and stimulate
metabolic and immune function. pituitary corticotrophs to release adrenocorticotropic
hormone (ACTH). ACTH is released into the systemic
biological rhythms are in synchrony. However, with our circulation and once at the adrenal cortex stimulates
increasingly chaotic lifestyles this orderly physiological the production of CORT. CORT undergoes de novo
regulation is steadily being disrupted, which can result synthesis and release back into the systemic circulation,
in chronodisruption4,10. This desynchronization between enabling it to travel to its target tissues and produce its
cellular oscillators in the SCN and peripheral tissues characteristic metabolic, cardiovascular, immunological
can manifest as negative health outcomes in the form and cognitive effects33. CORT also acts via an autoregu­
of cardiovascular, metabolic, cognitive and immune latory negative feedback loop and inhibits HPA activity
dysfunction4,10–14. via effects at the level of the pituitary, hypothalamic PVN
and hippocampus35.
HPA axis and circadian rhythmicity Further levels of circadian control exist, including
The HPA axis is critical for life and is a major part of splanchnic nerve innervation of the adrenal glands36.
our homeostatic regulatory system15. The output of this The adrenal glands receive autonomic (sympathetic)
system is the endogenous glucocorticoid corticosterone innervation via neuronal projections of the autonomic
(in rodents) or cortisol (in humans), which are collec­ portion of the PVN36,37, which alters adrenal cortical
tively referred to as CORT. Glucocorticoids have diverse sensitivity to ACTH with a reduction in responsive­
and far reaching effects, which is why they are such suc­ ness during the circadian nadir. CORT synthesis and
cessful therapeutic agents; however, this diversity is a adrenal clock gene functioning is also influenced by a
double-​edged sword and excess levels of glucocorticoids light-​sensitive mechanism that occurs across the 24-h
result in a myriad of unwanted adverse effects, including period, with a shift in irradiance threshold according to
diabetes mellitus, hypertension, immune dysregulation the time of day. Thus, in mice housed in complete dark­
and osteoporosis16. Glucocorticoids exhibit powerful ness, although high-​intensity light activated cortico­
anti-​inflammatory functions both at a whole-​cell and sterone at all times of day, lower intensity light had no
at a transcriptional level. They can induce apoptosis of effect during the subjective day. The adrenal response
T lymphocytes, neutrophils, basophils and eosinophils17. therefore is dependent on both irradiance and circadian
They also regulate multiple proinflammatory genes phase38,39. The adrenal gland itself also has an autono­
encoding cytokines, chemokines and inflammatory mous clock that regulates ACTH sensitivity and steroido­
enzymes associated with repression of AP1 and nuclear genesis, allowing it to fine tune its own homeostatic
factor-​κB (NF-​κB) transcription18. Glucocorticoids also control40,41. Finally, peripheral CLOCK-​mediated acetyl­
inhibit antigen presentation19,20, major histocompatibil­ ation of the glucocorticoid receptor can decrease tissue
ity complex class II expression21 and antibodies22, and sensitivity to glucocorticoids in a circadian manner42.
favour T helper 1 versus T helper 2 responses20. They These circadian fluctuations in activation of gluco­
influence cytotoxic effects via cell death and oxidative corticoid receptors also have important interactions
Indirect projections stress23, have a role in metabolic regulation through glu­ with multiple other crucial homeostatic processes,
Neural pathways involving cose utilization and ATP production24 and interact with including the transcriptional activity of other genes that
at least one relay.
the major neurotransmitters and many secondary neuro­ respond to glucocorticoids and their corresponding
Hypophyseal portal system peptidergic systems. As such, glucocorticoids modulate physiological outputs, such as physical activity and body
The microcirculation that emotion and cognition, with key examples being learn­ temperature5,43. For example, in the rat, glucocorticoid-​
allows transport of ing ability, performance, emotional perception and responsive tryptophan hydroxylase 2, a gene impli­
hypothalamic hormones mood25,26. These interactions also exemplify how gluco­ cated in physical activity, temperature and emotional
to the pituitary gland.
corticoid therapy can result in multiple effects, including response, has a circadian rhythmicity that is abolished
Irradiance threshold unwanted adverse effects such as depression27–30. by exogenous steroids44.
The threshold power of (solar) CORT is a homeostatic anticipatory hormone that
electromagnetic radiation is secreted by the adrenal glands. Consequently, under Stress response
needed to exert an effect.
basal conditions it is released with a characteristic cir­ Acute stress. The acute response to stress is a dyna­mic
Stereotypic behaviours cadian pattern of secretion with high levels just before process that changes over time, starting with stereotypic
Repetitive body movements waking (start of the active cycle), followed by a steady behaviours and then changing to goal-directed behaviours
that serve no biological decline down to trough (or nadir) levels during the sleep­ specific to the stressor, followed by activation of the
function. ing or inactive phase, hence anticipating the needs of the SAM within seconds and finally recruitment of the HPA
Goal-​directed behaviours
body (Fig. 2a). The daily rhythm of CORT is regulated axis, with peak levels of cortisol occurring between
Behaviours engaged for a through indirect projections from the SCN to the para­ 15 and 20 min after stress onset45. These early responses
specific functional purpose. ventricular nucleus (PVN) of the hypothalamus, which provide increased energy resources and initiate longer

www.nature.com/nrendo
Reviews

Chronic stress. In response to chronic stress, a dynamic


Light Dark Hypothalamus change in the ratio of AVP to CRH in the hypothalamic
PVN
Suprachiasmatic PVN occurs49 as well as an associated decreased sen­
nucleus sitivity to the glucocorticoid feedback50. Obstructive
sleep apnoea is a good example of chronic stress (Fig. 2c).
In sleep apnoea, there is a marked increase in the amount
of cortisol released during each secretory pulse, which
Neuron normalizes after continuous positive airway pressure
treatment51. In critical illness, the situation is somewhat
different with the increased levels of cortisol produced
by long-​term stress being present for the first few days
Cortisol
secondary to increased adrenal sensitivity to ACTH and
increased cortisol synthesis47. During long-​term critical
Adrenal illness, a further change in HPA axis regulation occurs
gland
with reduced cortisol metabolism becoming an increas­
ingly important factor in maintaining raised levels of
plasma cortisol52,53.
Anterior Posterior
pituitary pituitary Glucocorticoid signalling. Glucocorticoid receptors
gland gland (GRs) and mineralocorticoid receptors (MRs) are the
cognate intracellular nuclear receptors for CORT54.
The affinity of CORT for MR is approximately fivefold to
ACTH tenfold higher than that for GR55. Binding of CORT to its
receptors leads to either transactivation or repression of
genomic transcription as well as more rapid nongenomic
effects56,57. Nongenomic signalling is mediated via clas­
Other sic58 or membrane-​bound variants of the receptors59,60.
Immune Peripheral
homeostatic
function clocks The membrane-​bound variants have lower glucocor­
signals
ticoid affinity than their classic nuclear counterparts61.
Nuclear MR are generally constantly occupied during
the day and only become unoccupied at the very low
levels of CORT found at night in humans or during the
Cognition Cardiovascular Metabolism
day in rodents. As CORT levels rise to a critical threshold
(as seen during the circadian peak or following acute or
chronic stress), nuclear GR and membrane-​associated
MR and GR occupation occurs54. It is important to note
that the one exception to this effect is the hypothalamic
Fig. 1 | coordination of central and peripheral clocks by glucocorticoids. The
supra­chiasmatic nucleus central clock receives light–dark signals that, in turn, influence SCN, which does not appear to be regulated by circu­
hypothalamic–pituitary–adrenal and sympatho–adrenomedullary activity leading to lating CORT62. As such, the SCN is the one place where
circadian CORT production. CORT activates glucocorticoid receptors in peripheral tissues, endogenous CORT cannot shift clock function. Whether
which synchronizes peripheral clocks and downstream metabolic, cardiovascular, neuronal this feature is due to a lack of corticosteroid receptors or
and immune pathways. Other Zeitgebers such as food, temperature and social cues can also altered chromatin structure is unclear.
entrain or influence the entrainment of clocks and can alter the output of these downstream A further level of regulatory control is the tissue
pathways. ACTH, adrenocorticotropic hormone; PVN, paraventricular nucleus. specificity of GR and MR distribution. GR are present
throughout the brain and peripheral tissues while MR
term and slower genomic effects that restrain inflam­ have more limited localization, predominately being
matory and other potentially dangerous responses46. The found in cardiovascular tissue, liver and kidneys, as well
response to the acute stress of cardiac surgery can be as corticolimbic regions of the brain54. Although GR are
seen in Fig. 2b (ref.47). This response is very interesting present throughout the brain, only the hippocampus,
for several reasons. First, despite the greatly increased basal ganglia, lateral septum and medial amygdala neu­
levels of cortisol, the pattern of cortisol secretion rons present a high MR:GR ratio63. As these areas do not
remains pulsatile. Second, despite initial high levels of express 11β-​hydroxysteroid dehydrogenase (11β-​HSD)
ACTH, these rapidly fall to basal levels while the cor­ type 2 (discussed in detail in a subsequent paragraph),
tisol level remains raised. Despite this fall in ACTH MR are persistently occupied even during the circa­
levels, small changes in these basal levels of ACTH ini­ dian nadir and it is GR and the fast-​acting nongenomic
tiate large pulses of cortisol release, indicating a rapidly response of the lower affinity membrane-​bound MR
induced increased sensitivity of the adrenal cortex to that respond when CORT levels rise in response to a
ACTH. This effect has now been investigated in reverse stressor28. This response helps to prepare an individual
translation studies in rats and complemented with mathe­ to respond to a stressor through enhancing synaptic
matical modelling, which has enabled the importance plasticity at a cellular level, which leads to a behavioural
of the dynamic adrenal steroidogenic regulatory network change in the form of altered decision making, atten­
to be characterized48. tional bias and risk assessment64. Although other brain

Nature Reviews | Endocrinology


Reviews

a Normal conditions in a healthy volunteer c Chronic stressor: obstructive sleep apnoea


Mean circadian Ultradian
cortisol ACTH Cortisol 600 Baseline
60
50 500 600

Cortisol (nmol/l)

Serum levels of cortisol


ACTH (pg/ml)
40 400 500
30 300 400

(nmol/l)
20 200 300
10 100
200
0 0
100
0
0
0
0
0
0
0
0
0
0
0
0
0
:0
:0
:0
:0
:0
:0
:0
:0
:0
:0
:0
:0
:0
19
21
23
01
03
05
07
09
11
13
15
17
19
0
Time (24 h)

b Acute stressor: cardiac surgery


1,500 1,000 600

Serum levels of cortisol


1,250 500
800

Cortisol (nmol/l)
ACTH (pg/ml)

1,000 400

(nmol/l)
600
750 300
400 200
500
250 200 100
0 0 0
0 0 0 0 0 0 0 0
:0 2:2 1:4 6:0 9:2 1:4 5:0 8:2
4
4
4
4
4
4
4
4
4
4
4
4
4
19
:0
:0
:0
:0
:0
:0
:0
:0
:0
:0
:0
:0
:0
2 0 0 0 1 1 1
08
10
12
14
16
18
20
22
00
02
04
06
08
Time (24 h) Time (24 h)

Fig. 2 | Human cortisol ultradian rhythmicity under basal and stressful conditions. Cortisol is secreted in a circadian
rhythm characterized by a nadir or quiescent phase during the inactive period (overnight while sleeping in humans).
An anticipatory rise in CORT levels occurs before waking followed by the circadian peak of secretion at the start of
the active period. Underlying this rhythm, however, is a complex and dynamic ultradian rhythm consisting of pulses
approximately every 3 h. a | A schematic representation of a circadian and ultradian cortisol rhythm from a healthy male
volunteer. b | The effect of acute stress during cardiac surgery (the shaded area shows when coronary artery bypass
grafting occurred). Cortisol levels remain elevated throughout the sampling period. This occurs via an initial surge in
adrenocorticotropic hormone (ACTH), followed postoperatively by altered adrenal sensitivity to ACTH. c | The effect of
chronic stress on hypothalamic–pituitary–adrenal axis activity in the form of obstructive sleep apnoea. Here, disordered
cortisol pulsatility is seen in the form of an increase in the number of pulses and the amount of cortisol secreted per pulse
at baseline (before continuous positive airway pressure (CPAP) therapy), especially over the quiescent period when
endogenous cortisol levels naturally fall. The profiles improve after 3 months of CPAP therapy. Parts a and b are modified
with permission from Wolters Kluwer, Gibbison, B. et al. Dynamic pituitary–adrenal interactions in response to cardiac
surgery. Crit. Care Med. 43, 791–800 (2015)47. Part c is adapted with permission from ref.51, Oxford University Press.

areas can still respond to changes in CORT this will (and therefore biologically active) CORT during the cir­
only be through their genomic and nongenomic GR65. cadian peak, which accentuates the diurnal profile of free
Consequently, this region specificity in receptor distri­ CORT and the response to stress. CBG also acts as a car­
bution will modify corticosteroid signalling, including rier to specific target sites (such as sites of inflammation)
the activation and termination of stress responses, and is and even acts as a protein thermocouple releasing CORT
likely to be important for promoting long-​lasting adap­ at sites of increased temperature71. CBG concentrations
tive protective mechanisms such as strategic planning are therefore of great importance for the bioavailability
and memory storage and consolidation66. of CORT and its ability to act at a tissue level72.
Other layers of regulatory control arise through avail­ The metabolic clearance of CORT occurs through
ability of CORT. CORT is highly lipophilic, which means two mechanisms, enzymic degradation in the liver72,73
that under basal conditions 90% of CORT is tightly and intracellular metabolism by the two isoenzymes of
bound to the high-​affinity, limited-​capacity protein car­ 11β-​HSD, which catalyse the interconversion of active
rier corticosteroid-​binding globulin (CBG)67,68. Around CORT and the inert forms, cortisone (human) and
4–5% is loosely bound to albumin and the remaining 5% 11-dehydrocorticosterone (rodents). 11β-​HSD type 1 is
of CORT is ‘free’ and biologically active69. CBG levels are predominately a reductase that catalyses the regenera­
controlled by a variety of different factors such as genetic tion of active glucocorticoids, which amplifies the cel­
variability, liver and thyroid function, nutritional status, lular actions of CORT73. It is widely expressed in liver,
inflammation and stress. Additionally, CBG is saturated adipose tissue, muscle, pancreatic islets, adult brain,
at fairly low CORT concentrations, including under basal inflammatory cells and gonads74. 11β-​HSD type 1 there­
(nonstressed) levels found at the circadian peak. Similar fore increases tissue levels of CORT and is thought to be
to CORT, CBG exhibits homeostatic diurnal varia­tion70, a major factor in regulating tissue sensitivity to CORT,
the resultant effect being a higher proportion of free especially under conditions that can increase 11β-​HSD

www.nature.com/nrendo
Reviews

Hypothalamus loss of circadian rhythmicity, but the underlying pulsa­


tility of CORT remains79. Nor does the ultradian rhythm
arise as one would expect from a central hypothalamic
‘pulse generator’, as pulses of CRH do not induce down­
Anterior
pituitary stream pulses of ACTH and CORT80. Mathematical
Cortisol
gland modelling techniques have consequently been utilized,
which predict that the ultradian rhythm has a subhy­
Adrenal Posterior
gland pituitary pothalamic origin62. Here, a dynamic feedforward–
gland feedback communication exists between ACTH on
ACTH the adrenal cortex and endogenous glucocorticoids
on the pituitary corticotrophs, together driving ultradian
rhythmicity with a natural delay between ACTH acti­
vating adrenal melanocortin 2 receptors, glucocorticoid
Adrenal delay biosynthesis and cortisol release (Fig. 3). This prediction
Cholesterol was subsequently confirmed in our rat model62.
MC2R
The adrenal gland itself preferentially responds to
oscillatory ACTH signals; in rodents whose endoge­
nous ACTH is suppressed with exogenous glucocor­
ticoids, pulsatile ACTH infusions result in pulsatile
corticosterone secretion whereas constant ACTH infu­
sions of the same total dose result in no response81.
Mitochondrion Additionally, the adrenal gland has its own internal feed­
forward–feedback system that amplifies the response
to rapid changes in ACTH level, sensitizing the adre­
nal glands to oscillating levels of ACTH and hence
alterations in CORT82.
Fig. 3 | Schematic representation of the hypothalamic–pituitary–adrenal axis
showing natural inbuilt adrenal delays. After release from the pituitary gland, Effect of hormone oscillation. Both circadian and ultra­
adrenocorticotropic hormone (ACTH) binds to adrenal melanocortin 2 receptors dian oscillations in hormone levels matter. Between
(MC2Rs). Here, natural inbuilt delays occur ; MC2R binding leads to increased uptake 5% and 20% of cells show clear circadian oscillations in
of cholesterol, which is the precursor for all steroid hormones. Cholesterol then mRNA levels12 and circadian fluctuations are found at
undergoes a series of biosynthetic steps before being transported into mitochondria, almost every stage of gene expression83 and have been
undergoing hydroxylation and the subsequent release of cortisol into the circulation. associated with important biological effects. In trans­
genic mice, if circadian peak levels of CORT are admin­
type 1 activity such as visceral obesity72. In addition, istered during a motor learning task, these mice show
11β-​HSD type 1 is thought to mediate many cognitive increased memory of motor skills84. This effect arises sec­
and metabolic effects of CORT excess. For instance, ondary to a rapid GR-​mediated nongenomic mechanism
11β-​HSD type 1 deficiency in rodents protects against via LIM kinase signalling that increases numbers of new
age-​related and stress-​related cognitive decline while neuronal spines. Low trough levels of CORT are then
transgenic overexpression exacerbates cognitive impair­ needed to retain and stabilize the memory. This reten­
ment75. The other isoform, 11β-​HSD type 2, is a high-​ tion and stabilization involves ‘pruning’ of old spines via
affinity dehydrogenase that inactivates glucocorticoids an MR-​mediated pathway that alters gene expression,
and is particularly important in the kidney, where it which implies that circadian fluctuations are required
protects mineralocorticoid receptors from activation for plasticity in learning and memory84. Indeed, clini­
by the much higher concentration of cortisol73. More cally this phenomenon is nothing new; in patients tak­
studies are needed to define how tissue ratios of cortisol ing glucocorticoid-​based therapeutics and in those with
and cortisone might change during stress and whether Cushing syndrome, both situations with chronically ele­
this might provide yet another mechanism to guard our vated glucocorticoid levels and/or no oscillations, there
homeostatic state. are marked impairments in learning and memory25.
What we are beginning to understand is that circa­
Ultradian rhythm. Underlying the circadian rhythm there dian rhythmicity is only the tip of the iceberg, and func­
is a complex and dynamic ultradian rhythm comprised tionally important dynamic changes are seen in a shorter
of discrete pulses of secretion of both ACTH and cor­ time frame than can be described by simple circadian
tisol (Fig. 2a). This pulsatile pattern has an approximate alterations. In the HPA axis, ultradian hormone pres­
frequency of 60–90 min with increased pulse amplitude entation is essential for physiological gene functioning85.
and frequency at the circadian peak76. Patterns of secre­ Very different patterns of gene activation are seen both
tion of ACTH and cortisol are influenced by a multitude in vitro and in vivo depending on the pattern of hormone
of factors, including genetics, sex hormones, epigenetic presentation86. Ultradian oscillations in glucocorticoid
influences, environmental stressors and age, which leads levels result in a phenomenon known as gene pulsing
Circadian rhythm
Any biological process that
to individual diversity77,78. Unlike circadian rhythms, the (Fig. 4a). This phenomenon is specific for the endoge­
displays an oscillation of origin of the ultradian rhythm is not from central SCN nous glucocorticoid hormones as synthetic glucocorti­
approximately 24 h. regulation; rodents with SCN lesions show an isolated coids have long binding characteristics at the GR and

Nature Reviews | Endocrinology


Reviews

cannot cycle on and off86. As CORT levels rise, as is seen in pulse termination, GR dissociates from DNA and
in endogenous glucocorticoid pulsatility, GR binding is released back into the nucleoplasm ready to rapidly
and activation occurs followed by nuclear translocation, respond to subsequent CORT pulses. If the same dose of
dimerization and interaction with DNA glucocorticoid CORT is used but is presented as a constant as opposed
response elements, which initiates transcription. Next, to a pulsatile pattern of presentation, genomic reactivity
rapid cycling of GR and transcription factors on and off changes lead to altered transcriptional activity86,87 as well
chromatin occurs. Once CORT levels decline, as seen as differential gene responses86 (Fig. 4b). In addition to

a Gene pulsing b Cell culture (Tsc22d3)


Pulses of corticosterone 25 Pulsed Constant
GR–DNA binding
Per1 nascent RNA
15.0 20
1,500
12.5
corticosterone (ng/ml)

Fold change
1,250 15
Plasma levels of

Fold induction
10.0
1,000

750 7.5 10

500 5.0
5
250 2.5

0 0 0
0 30 60 90 120 150 180 C P1 W1 P2 W2 P3 W3 P4 W4 P5 W5 P6 W6 P7 W7 P8 W8
Time (min)

c Animal model — rodent amygdala d Human model — BOLD fMRI responses to FERT
P < 0.05 Nonpulsatile Pulsatile glucocorticoid Attentional bias towards
glucocorticoid rhythm rhythm emotional faces
30 50 P = 0.014
P < 0.01

Vigilance scores
Grey values (arb. units x104)

25
P < 0.01

20

15 Striatum
Insula

y
10
rfu

rfu
pp

pp
a

a
Ha

Ha
Fe

Fe
5
Nonpulsatile Pulsatile
0
Recognition of emotional faces
Vehicle 80
Amygdala
Constant
Accuracy (%)

P = 0.01

50 ng/ml corticosterone Uncertainty in identifying


emotional valence
50 ng/ml corticosterone

100 ng/ml corticosterone ve tiv


e ve tiv
e
siti ga siti ga
Po Ne Po Ne
100 ng/ml corticosterone
Nonpulsatile Pulsatile

Fig. 4 | The importance of gene pulsing. a | In adrenalectomized rats, depending on the pattern of exposure. d | Healthy human male volunteers
hourly pulses of corticosterone result in a similar pattern of association– underwent a chemical adrenalectomy with metyrapone. Data from
dissociation kinetics in glucocorticoid receptor (GR)–DNA binding and ref. 93. In a cross-​over trial design they then received hydrocortisone
downstream cyclic recruitment and pulsatile release of the clock gene Per1 replacement therapy (total daily dose 20 mg) as either standard oral
nascent RNA. This effect is not seen with synthetic glucocorticoid (three times a day), constant circadian subcutaneous infusion or constant
treatment. b | Ultradian and constant corticosterone treatments have pulsatile subcutaneous infusion for 5 days. On day 5, participants
different effects on transcription of Tsc22d3, a GR-​regulated gene. Here, underwent functional MRI (fMRI) while performing a facial expression
pulsatile presentation shows a pulsatile pattern of gene fold induction, recognition task (FERT). Blood-​oxygen-level-​dependent (BOLD) imaging
while constant infusion induces constant RNA release. c | Adrenalectomized showed a differential response in attentional bias and recognition of
rats received either pulsatile corticosterone (50 ng/ml or 100 ng/ml peaks) emotional faces depending on whether a pulsatile or a nonpulsatile
or constant corticosterone (12-h constant infusion clamped at 50 ng/ml). pattern of glucocorticoid presentation was used. Communication
They then underwent a 10-min noise stress. c-​fos mRNA expression was networks within the insula, striatum and amygdala also varied depending
analysed in different brain regions. A differential response was found in the on whether the presentation was pulsatile or nonpulsatile. C, control;
pituitary , amygdala, hippocampus and paraventricular nucleus. The effect P, induction; W, withdrawal. Parts a and b are reproduced from ref. 86,
shown was dependent on both phase and amplitude of the glucocorticoid Springer Nature Limited. Part c is adapted with permission from ref.91,
pulse, which suggests that a differential response is seen in stress circuitry Oxford University Press.

www.nature.com/nrendo
Reviews

the classic glucocorticoid response element-​dependent making and alertness and stimulates cognitive function­
interactions, GR can also interact with a variety of other ing. CORT also has a proinflammatory effect, resulting
transcription factors and influence transcription via in an increased response to infection99. Chronic expo­
protein–protein interactions88,89 and chromatin remodel­ sure to stress results in reversal of the beneficial effects
ling88,90. These pattern-​dependent interactions have seri­ with long-​term cortisol exposure becoming maladaptive,
ous implications for therapeutics based on continuous which leads to a broad range of symptoms and disease
exposure to glucocorticoids, and probably contribute to states, including the metabolic syndrome, obesity, can­
the adverse effects of these drugs16. cer, mental health disorders, cardiovascular disease and
Downstream, constant and pulsatile patterns of increased susceptibility to infections10,11,54,100,101.
CORT result in differential neuronal and behavioural With regard to immune functioning, components of
responses. For instance, noise stress induces neuronal both the adaptive and innate immune system undergo cir­
activation of c-​fos in the amygdala, hippocampus and cadian variations in levels and functionality to optimize
hypothalamic PVN. Interestingly, the pulse phase is cru­ response and recovery to a pathogen102. Autonomous
cial for this response. If the noise is administered during circadian clocks are also found in different subsets of
the rising phase, the c-​fos response in the amygdala is immune cells, including macrophages and lympho­
increased (Fig. 4c). This effect is paralleled by differen­ cytes103,104. CORT also has a multitude of effects on the
tial behavioural responses to the stressor. Importantly, immune system20. Chronic increases in CORT, either as
the effect is reproducible with the pulse phase depend­ a result of the effect of the steroid or the development of
ency pattern being recorded in aggressive and novelty glucocorticoid resistance101, increases susceptibility to
behaviour responses to glucocorticoids, as well as in infection20. Chronic stress can impair GR function as a
the neuroendocrine mechanism of rapid HPA negative result of long-​term exposure to the resultant release of
feedback91,92. Translational studies in humans have found proinflammatory cytokines. Indeed, cytokines (along with
similar results (Fig. 4d). Healthy male volunteers on a their signalling pathways including, but not limited to,
block and replace regimen of endogenous glucocorticoid mitogen-​activated protein kinases, NF-​κB and cycloox­
blockade with metyrapone and replacement therapy ygenase) inhibit GR function99. Potential mechanisms
with either circadian or ultradian subcutaneous hydro­ underlying this inhibition include disruption of GR trans­
cortisone or standard oral hydrocortisone therapy (three location, GR–DNA binding, GR–inflammatory media­
times a day) at the same total daily dose exhibit altered tor interactions and alterations in GR phosphorylation
cognitive and behavioural effects that are dependent on status105–107. Glucocorticoid resistance in turn interferes
the cortisol pattern. Specifically, a lack of physiological with HPA downregulation of the very same proinflam­
pulsatility was associated with poorer quality of sleep, matory cytokines and creates a vicious cycle. Thus, in
poorer working memory performance and altered accu­ the case of the common cold, an exaggerated response to the
racy in recognition and attentional bias toward or away symptoms and signs of the upper respiratory tract illness
from emotional faces93. is generated by the resultant proinflammatory response108.
Long-​term exposure to CORT also increases blood
Stress and disease levels of glucose109 and stimulates adipocyte precursor
Stress is any stimulus that disrupts or threatens homeo­ maturation, which promotes adiposity110. Adipose tissue
static balance. The stress response is a mechanism that is now seen as an active endocrine gland rather than a
can restore homeostatic processes and promote self-​ passive energy store as it produces a wide variety of hor­
preservation through a complex interaction between mones and regulatory factors72. The levels of these fac­
the HPA axis, central and peripheral autonomic nervous tors in turn reflect the metabolic status of the adipocytes
systems and immune systems15. The HPA axis responds and interact with homeostatic connections between adi­
to stress by rapidly releasing CORT. As CORT is highly pose tissue and the brain as well as other organs, which
lipophilic, it is not prestored in vesicles and is synthesized together control energy intake and expenditure72,111.
de novo in response to ACTH. During long-​term stress a Obesity is also linked to chronic low-​grade inflamma­
dissociation between ACTH and CORT secretion deve­ tion108 via tumour necrosis factor (TNF) and IL-6. TNF
lops94,95, predominantly via increased adrenal sensitivity inhibits intracellular insulin signalling pathways and
to ACTH48 or non-​ACTH-mediated mechanisms95. This IL-6 promotes atherosclerosis and metabolism of glucose
effect is seen in patients undergoing cardiac surgery, who and lipids111,112, all of which interact with the HPA axis20.
show a marked shift in adrenal responsiveness47 (Fig. 2b). Studies of an increasingly common stress-​associated
Reverse translation studies in animals suggest this shift problem — sleep deprivation — confirm elevated nadir
arises secondary to changes in the regulation of both stim­ CORT levels, which then acts as another risk factor for
ulatory and inhibitory intra-​adrenal signalling pathways47. insulin resistance113. Behaviourally, chronic sleep depri­
In models of inflammatory bowel disease and viral chal­ vation is associated with increased appetite and energy
lenges, IL-6 and ACTH-​independent immune–adrenal expenditure114. Chronic CORT exposure also stimulates
pathways have also been linked to this dissociation96,97. mesolimbic reward pathways within the brain, leading
The rapid rise in levels of CORT seen under condi­ to increased intake of palatable food such as sucrose
tions of stress increases the availability of supplemen­ solutions111. Additionally, increased feeding might be
tal energy stores by promotion of gluconeogenesis and stimulated by leptin and ghrelin levels115,116. During
inhibition of insulin production combined with vaso­ chronic stress, it is thought that these two hormones can
constriction, which aids delivery of blood to the mus­ no longer accurately signal caloric need117. The effect in
cles and the brain98. Centrally, CORT promotes decision this situation is a misperception of insufficient available

Nature Reviews | Endocrinology


Reviews

energy stores116. This effect is seen in combination with from fatigue and difficulties in concentrating130–132. This
increased proinflammatory cytokine levels28, which therapy fails to provide the anticipatory rise in cortisol
makes it is unsurprising that chronic stress and chrono­ before wakening and thus gives a shifted circadian pro­
disruption are linked with the development of metabolic file while also resulting in acute nonphysiological lev­
complications. els of cortisol in the middle of the day133. Patients have
Hypercortisolaemia is also linked with affective dis­ altered immune responses in the form of altered circa­
orders10,27. Mathematical modelling techniques have dian patterns of circulating monocytes and natural killer
shown that individuals with major depressive disorder cells134, impaired natural killer cell function135 and hypo­
have disordered cortisol secretion with an increased methylated CD4+ T cells136. In an attempt to rectify these
mass of cortisol in each pulse and increased pulse fre­ adverse outcomes, modified and dual-​release prepara­
quency118. Patients with major depressive disorder have tions134,137,138, as well as continuous139 and pulsatile pump
long been known to exhibit glucocorticoid resistance (The Pulses Study; ISRCTN67193733) preparations, are
in the form of resistance of endogenous cortisol to sup­ being trialled. Initial results are encouraging, with closer
pression in the dexamethasone and dexamethasone– mirroring of circadian profiling resulting in alterations
CRH suppression tests119, the results of which can pre­ in clock gene function140 and improvements in immune
dict clinical outcome120. Chronic exposure to stress functioning134 and quality of life134,137,141,142. These issues
results in a degree of limbic system atrophy, a key region are not only limited to endogenous CORT, but also to
involved in regulation of emotion 121. This effect is synthetic glucocorticoids16. These are some of the most
also closely interlinked with immune function; stress-​ widely prescribed medications143,144 but, despite their
induced proinflammatory cytokines, including IL-1β, clinical efficacy, over three quarters of patients experi­
IL-6 and TNF, are implicated in depressive behaviour ence adverse effects even at what would be considered
in rodent models of depression and in patients with a fairly low dose145,146. Chronic glucocorticoid treatment
depression101,105,122,123. The mechanisms underlying this can also induce glucocorticoid resistance in susceptible
connection are complex. It is possible that inflamma­ patients with glucocorticoid-​responsive conditions such
tion and glucocorticoid signalling might act on the same as asthma or rheumatoid arthritis147.
processes to cause cumulative damage or that the dual
pro­inflammatory and anti-​inflammatory effects of gluco­ Conclusions
corticoids disrupt the immune system of the brain Homeostatic processes are dynamic and interactive.
in favour of inflammatory effects124. In patients with As society changes with the development of a 24-h world
depression, decreased T cell availability might arise sec­ and its use of social media and changes in interpersonal
ondary to increased apoptosis sensitivity combined with communication, many of our evolutionarily adaptive
impaired glucocorticoid responsiveness and reduced processes are in danger of becoming maladaptive. If we
brain trafficking capacity in response to the appro­ are to understand the growing epidemic of stress-​related
priate neuroendocrine or immune stimuli101,122. Stress human disease, we need to go back to first principles
also enhances NF-​κB activation106,107,125; indeed, stress-​ and understand not only the mechanisms underlying the
induced anhedonia is directly linked to increased NF-​κB regulation of our patterns of physiological activity and
activity105. Interestingly, antidepressants can enhance GR hormone secretion — particularly over the important
functioning126 and inhibit cytokine pathways127. and notoriously difficult nadir period during sleep —
These effects of glucocorticoids are usually accepted but also why they are important for the maintenance of
as simply being due to the high levels found in the blood. health. If we are going to aspire to an objective of opti­
However, this theory might be an oversimplification. mal personalized medicine, we need to devise methods
In primary hypocortisolaemia (or Addison disease), not only to measure dynamic basal patterns of hor­
patients are treated with what we believe to be opti­ monal metabolic and immune functioning over the
mal replacement of hydrocortisone three times a day whole day, but also novel therapeutic interventions to
to mimic the circadian rhythm128,129. Despite this effort to counteract the causes of environmental-​related and/or
give physiological replacement, these patients have stress-​related illness.
increased mortality as a result of cardiovascular disease,
infectious disease and cancer, and increased morbidity Published online xx xx xxxx

1. Szabo, S., Tache, Y. & Somogyi, A. The legacy of Hans 7. Skene, D. J. et al. Separation of circadian- and in mammals: implications for biology and medicine.
Selye and the origins of stress research: a retrospective behavior-​driven metabolite rhythms in humans Proc. Natl Acad. Sci. USA 111, 16219–16224
75 years after his landmark brief “letter” to the editor provides a window on peripheral oscillators and (2014).
of Nature. Stress 15, 472–478 (2012). metabolism. Proc. Natl Acad. Sci. USA 115, 13. Smarr, B. L. & Schirmer, A. E. 3.4 million real-​world
2. Levine, S. Influence of psychological variables on the 7825–7830 (2018). learning management system logins reveal the
activity of the hypothalamic-​pituitary-adrenal axis. 8. Buhr, E. D. & Takahashi, J. S. Molecular components majority of students experience social jet lag
Eur. J. Pharmacol. 405, 149–160 (2000). of the mammalian circadian clock. Handb. Exp. correlated with decreased performance. Sci. Rep. 8,
3. Brown, S. A. & Azzi, A. Peripheral circadian oscillators Pharmacol. 217, 3–27 (2013). 4793 (2018).
in mammals. Handb. Exp. Pharmacol. 2013, 45–66 9. Takahashi, J. S. Transcriptional architecture of the 14. Gardner, M. et al. Dysregulation of the hypothalamic
(2013). mammalian circadian clock. Nat. Rev. Genet. 18, pituitary adrenal (HPA) axis and cognitive capability at
4. Roenneberg, T. & Merrow, M. The circadian clock 164–179 (2017). older ages: individual participant meta-​analysis of five
and human health. Curr. Biol. 26, R432–R443 10. Baron, K. G. & Reid, K. J. Circadian misalignment and cohorts. Sci. Rep. 9, 4555 (2019).
(2016). health. Int. Rev. Psychiatry 26, 139–154 (2014). 15. Selye, H. Stress and the general adaptation syndrome.
5. Bass, J. & Lazar, M. A. Circadian time signatures of 11. Potter, G. D. et al. Circadian rhythm and sleep BMJ 1, 1383–1392 (1950).
fitness and disease. Science 354, 994–999 (2016). disruption: causes, metabolic consequences, and 16. Russell, G. M. & Lightman, S. L. Can side effects of
6. Turek, F. W. Circadian neural rhythms in countermeasures. Endocr. Rev. 37, 584–608 (2016). steroid treatments be minimized by the temporal
mammals. Annu. Rev. Physiol. 47, 49–64 12. Zhang, R., Lahens, N. F., Ballance, H. I., Hughes, M. E. aspects of delivery method? Expert Opin. Drug Saf.
(1985). & Hogenesch, J. B. A circadian gene expression atlas 13, 1501–1513 (2014).

www.nature.com/nrendo
Reviews

17. Sorrells, S. F. & Sapolsky, R. M. An inflammatory 40. Ishida, A. et al. Light activates the adrenal gland: pathway: about receptors, transcription machinery
review of glucocorticoid actions in the CNS. timing of gene expression and glucocorticoid release. and microRNA. Brain Res. 1293, 129–141 (2009).
Brain Behav. Immun. 21, 259–272 (2007). Cell Metab. 2, 297–307 (2005). 66. Russell, G. M., Kalafatakis, K. & Lightman, S. L.
18. Busillo, J. M. & Cidlowski, J. A. The five Rs 41. Oster, H. et al. The circadian rhythm of glucocorticoids The importance of biological oscillators for HPA
of glucocorticoid action during inflammation: is regulated by a gating mechanism residing in the activity and tissue glucocorticoid response:
ready, reinforce, repress, resolve, and restore. adrenal cortical clock. Cell Metab. 4, 163–173 (2006). coordinating stress and neurobehavioural adaptation.
Trends Endocrinol. Metab. 24, 109–119 (2013). 42. Charmandari, E. et al. Peripheral CLOCK regulates J. Neuroendocrinol. 27, 378–388 (2015).
19. McEwen, B. S. et al. The role of adrenocorticoids as target-​tissue glucocorticoid receptor transcriptional 67. Lewis, J. G. et al. Plasma variation of corticosteroid-​
modulators of immune function in health and disease: activity in a circadian fashion in man. PLOS ONE 6, binding globulin and sex hormone-​binding globulin.
neural, endocrine and immune interactions. Brain Res. e25612 (2011). Horm. Metab. Res. 38, 241–245 (2006).
Rev. 23, 79–133 (1997). 43. Bailey, S. L. & Heitkemper, M. M. Circadian rhythmicity 68. Lewis, J. G., Bagley, C. J., Elder, P. A., Bachmann, A. W.
20. Elenkov, I. J. & Chrousos, G. P. Stress system — of cortisol and body temperature: morningness-​ & Torpy, D. J. Plasma free cortisol fraction reflects
organization, physiology and immunoregulation. eveningness effects. Chronobiol. Int. 18, 249–261 levels of functioning corticosteroid-​binding globulin.
Neuroimmunomodulation 13, 257–267 (2006). (2001). Clin. Chim. Acta 359, 189–194 (2005).
21. Brinkmann, V. & Kristofic, C. Regulation by 44. Donner, N. C., Montoya, C. D., Lukkes, J. L. 69. Hammond, G. L., Smith, C. L. & Underhill, D. A.
corticosteroids of Th1 and Th2 cytokine production in & Lowry, C. A. Chronic non-​invasive corticosterone Molecular studies of corticosteroid binding globulin
human CD4+ effector T cells generated from CD45RO- administration abolishes the diurnal pattern of tph2 structure, biosynthesis and function. J. Steroid
and CD45RO+ subsets. J. Immunol. 155, 3322–3328 expression. Psychoneuroendocrinology 37, 645–661 Biochem. Mol. Biol. 40, 755–762 (1991).
(1995). (2012). 70. Frairia, R. et al. Influence of naturally occurring and
22. Wiegers, G. J. & Reul, J. M. Induction of cytokine 45. Lightman, S. L. The neuroendocrinology of stress: synthetic glucocorticoids on corticosteroid-​binding
receptors by glucocorticoids: functional and a never ending story. J. Neuroendocrinol. 20, globulin-​steroid interaction in human peripheral
pathological significance. Trends Pharmacol. Sci. 19, 880–884 (2008). plasma. Ann. NY Acad. Sci. 538, 287–303 (1988).
317–321 (1998). 46. Nicolaides, N. C., Kyratzi, E., Lamprokostopoulou, A., 71. Cameron, A. et al. Temperature-​responsive release
23. Abraham, I. M., Meerlo, P. & Luiten, P. G. Chrousos, G. P. & Charmandari, E. Stress, the stress of cortisol from its binding globulin: a protein
Concentration dependent actions of glucocorticoids system and the role of glucocorticoids. thermocouple. J. Clin. Endocrinol. Metab. 95,
on neuronal viability and survival. Dose Response 4, Neuroimmunomodulation 22, 6–19 (2015). 4689–4695 (2010).
38–54 (2006). 47. Gibbison, B. et al. Dynamic pituitary-​adrenal interactions 72. Kyrou, I., Chrousos, G. P. & Tsigos, C. Stress, visceral
24. Plaschke, K., Muller, D. & Hoyer, S. Effect of in response to cardiac surgery. Crit. Care Med. 43, obesity, and metabolic complications. Ann. NY Acad.
adrenalectomy and corticosterone substitution 791–800 (2015). Sci. 1083, 77–110 (2006).
on glucose and glycogen metabolism in rat brain. 48. Spiga, F. et al. Dynamic responses of the adrenal 73. Chapman, K., Holmes, M. & Seckl, J. 11beta-​
J. Neural Transm. 103, 89–100 (1996). steroidogenic regulatory network. Proc. Natl Acad. hydroxysteroid dehydrogenases: intracellular gate-​
25. Belanoff, J. K., Gross, K., Yager, A. & Schatzberg, A. F. Sci. USA 114, E6466–E6474 (2017). keepers of tissue glucocorticoid action. Physiol. Rev.
Corticosteroids and cognition. J. Psychiatr. Res. 35, 49. Ma, X. M., Levy, A. & Lightman, S. L. Emergence of an 93, 1139–1206 (2013).
127–145 (2001). isolated arginine vasopressin (AVP) response to stress 74. Seckl, J. R. 11beta-​hydroxysteroid dehydrogenases:
26. Roozendaal, B. Stress and memory: opposing effects after repeated restraint: a study of both AVP and changing glucocorticoid action. Curr. Opin. Pharmacol.
of glucocorticoids on memory consolidation and corticotropin-​releasing hormone messenger ribonucleic 4, 597–602 (2004).
memory retrieval. Neurobiol. Learn. Mem. 78, acid (RNA) and heteronuclear RNA. Endocrinology 75. Verma, M. et al. 11beta-​hydroxysteroid
578–595 (2002). 138, 4351–4357 (1997). dehydrogenase-1 deficiency alters brain energy
27. Brown, E. S. Effects of glucocorticoids on mood, 50. Dallman, M. F. Stress update: adaptation of the metabolism in acute systemic inflammation. Brain
memory, and the hippocampus. Treatment and hypothalamic-​pituitary-adrenal axis to chronic stress. Behav. Immun. 69, 223–234 (2018).
preventive therapy. Ann. NY Acad. Sci. 1179, 41–55 Trends. Endocrinol. Metab. 4, 62–69 (1993). 76. Follenius, M., Simon, C., Brandenberger, G. & Lenzi, P.
(2009). 51. Henley, D. E. et al. Hypothalamic-​pituitary-adrenal Ultradian plasma corticotropin and cortisol rhythms:
28. de Kloet, E. R., Oitzl, M. S. & Joels, M. Stress and axis activation in obstructive sleep apnea: the effect time-​series analyses. J. Endocrinol. Invest. 10,
cognition: are corticosteroids good or bad guys? of continuous positive airway pressure therapy. 261–266 (1987).
Trends Neurosci. 22, 422–426 (1999). J. Clin. Endocrinol. Metab. 94, 4234–4242 (2009). 77. Hartmann, A., Veldhuis, J. D., Deuschle, M.,
29. Decani, S., Federighi, V., Baruzzi, E., Sardella, A. 52. Boonen, E. et al. Reduced cortisol metabolism during Standhardt, H. & Heuser, I. Twenty-​four hour cortisol
& Lodi, G. Iatrogenic Cushing’s syndrome and topical critical illness. N. Engl. J. Med. 368, 1477–1488 release profiles in patients with Alzheimer’s and
steroid therapy: case series and review of the literature. (2013). Parkinson’s disease compared to normal controls:
J. Dermatolog. Treat. 25, 495–500 (2014). 53. Peeters, B. et al. Adrenocortical function during ultradian secretory pulsatility and diurnal variation.
30. Kenna, H. A., Poon, A. W., de los Angeles, C. P. prolonged critical illness and beyond: a prospective Neurobiol. Aging 18, 285–289 (1997).
& Koran, L. M. Psychiatric complications of treatment observational study. Intensive Care Med. 44, 78. Rivest, R. W., Schulz, P., Lustenberger, S.
with corticosteroids: review with case report. 1720–1729 (2018). & Sizonenko, P. C. Differences between circadian
Psychiatry Clin. Neurosci. 65, 549–560 (2011). 54. De Kloet, E. R., Vreugdenhil, E., Oitzl, M. S. & Joels, M. and ultradian organization of cortisol and melatonin
31. Buijs, R. M., Markman, M., Nunes-​Cardoso, B., Brain corticosteroid receptor balance in health and rhythms during activity and rest. J. Clin. Endocrinol.
Hou, Y. X. & Shinn, S. Projections of the suprachiasmatic disease. Endocr. Rev. 19, 269–301 (1998). Metab. 68, 721–729 (1989).
nucleus to stress-​related areas in the rat hypothalamus: 55. Reul, J. M. & de Kloet, E. R. Two receptor systems for 79. Waite, E. J. et al. Ultradian corticosterone secretion is
a light and electron microscopic study. J. Comp. Neurol. corticosterone in rat brain: microdistribution and maintained in the absence of circadian cues. Eur. J.
335, 42–54 (1993). differential occupation. Endocrinology 117, 2505–2511 Neurosci. 36, 3142–3150 (2012).
32. Watts, A. G. & Swanson, L. W. Efferent projections (1985). 80. Ixart, G., Barbanel, G., Nouguier-​Soule, J.
of the suprachiasmatic nucleus: II. Studies using 56. Dallman, M. F. Fast glucocorticoid actions on brain: & Assenmacher, I. A quantitative study of the pulsatile
retrograde transport of fluorescent dyes and back to the future. Front. Neuroendocrinol. 26, parameters of CRH-41 secretion in unanesthetized
simultaneous peptide immunohistochemistry in the 103–108 (2005). free-​moving rats. Exp. Brain Res. 87, 153–158
rat. J. Comp. Neurol. 258, 230–252 (1987). 57. Russell, G. M. et al. Rapid glucocorticoid receptor-​ (1991).
33. Jacobson, L. Hypothalamic-​pituitary-adrenocortical mediated inhibition of hypothalamic-​pituitary-adrenal 81. Spiga, F. et al. ACTH-​dependent ultradian rhythm of
axis regulation. Endocrinol. Metab. Clin. North Am. ultradian activity in healthy males. J. Neurosci. 30, corticosterone secretion. Endocrinology 152,
34, 271–292 (2005). 6106–6115 (2010). 1448–1457 (2011).
34. Herman, J. P., Ostrander, M. M., Mueller, N. K. 58. Lowenberg, M., Verhaar, A. P., van den Brink, G. R. 82. Spiga, F., Liu, Y., Aguilera, G. & Lightman, S. L.
& Figueiredo, H. Limbic system mechanisms of stress & Hommes, D. W. Glucocorticoid signaling: a Temporal effect of adrenocorticotrophic hormone on
regulation: hypothalamo-​pituitary-adrenocortical axis. nongenomic mechanism for T cell immunosuppression. adrenal glucocorticoid steroidogenesis: involvement
Prog. Neuropsychopharmacol. Biol. Psychiatry 29, Trends Mol. Med. 13, 158–163 (2007). of the transducer of regulated cyclic AMP-​response
1201–1213 (2005). 59. Orchinik, M., Murray, T. F., Franklin, P. H. & Moore, F. L. element-​binding protein activity. J. Neuroendocrinol.
35. Dallman, M. F. et al. Corticosteroids and the control Guanyl nucleotides modulate binding to steroid 23, 136–142 (2011).
of function in the hypothalamo-​pituitary-adrenal receptors in neuronal membranes. Proc. Natl Acad. Sci. 83. Lim, C. & Allada, R. Emerging roles for post-​
(HPA) axis. Ann. NY Acad. Sci. 746, 22–31 (1994). USA 89, 3830–3834 (1992). transcriptional regulation in circadian clocks.
36. Jasper, M. S. & Engeland, W. C. Splanchnic neural 60. Orchinik, M., Murray, T. F. & Moore, F. L. A Nat. Neurosci. 16, 1544–1550 (2013).
activity modulates ultradian and circadian rhythms in corticosteroid receptor in neuronal membranes. 84. Liston, C. et al. Circadian glucocorticoid oscillations
adrenocortical secretion in awake rats. Science 252, 1848–1851 (1991). promote learning-​dependent synapse formation and
Neuroendocrinology 59, 97–109 (1994). 61. Joels, M., Pasricha, N. & Karst, H. The interplay maintenance. Nat. Neurosci. 16, 698–705 (2013).
37. Buijs, R. M. et al. Anatomical and functional between rapid and slow corticosteroid actions in brain. 85. Lightman, S. L. & Conway-​Campbell, B. L. The crucial
demonstration of a multisynaptic suprachiasmatic Eur. J. Pharmacol. 719, 44–52 (2013). role of pulsatile activity of the HPA axis for continuous
nucleus adrenal (cortex) pathway. Eur. J. Neurosci. 11, 62. Walker, J. J. et al. The origin of glucocorticoid hormone dynamic equilibration. Nat. Rev. Neurosci. 11,
1535–1544 (1999). oscillations. PLOS Biol. 10, e1001341 (2012). 710–718 (2010).
38. Kiessling, S., Sollars, P. J. & Pickard, G. E. 63. Patel, P. D. et al. Glucocorticoid and mineralocorticoid 86. Stavreva, D. A. et al. Ultradian hormone stimulation
Light stimulates the mouse adrenal through a receptor mRNA expression in squirrel monkey brain. induces glucocorticoid receptor-​mediated pulses of gene
retinohypothalamic pathway independent of an effect J. Psychiatr. Res. 34, 383–392 (2000). transcription. Nat. Cell Biol. 11, 1093–1102 (2009).
on the clock in the suprachiasmatic nucleus. PLOS ONE 64. Groeneweg, F. L., Karst, H., de Kloet, E. R. & Joels, M. 87. Conway-​Campbell, B. L., Pooley, J. R., Hager, G. L.
9, e92959 (2014). Rapid non-​genomic effects of corticosteroids and their & Lightman, S. L. Molecular dynamics of ultradian
39. Husse, J., Leliavski, A., Tsang, A. H., Oster, H. role in the central stress response. J. Endocrinol. 209, glucocorticoid receptor action. Mol. Cell. Endocrinol.
& Eichele, G. The light-​dark cycle controls peripheral 153–167 (2011). 348, 383–393 (2012).
rhythmicity in mice with a genetically ablated 65. de Kloet, E. R., Fitzsimons, C. P., Datson, N. A., 88. George, C. L., Lightman, S. L. & Biddie, S. C.
suprachiasmatic nucleus clock. FASEB J. 28, Meijer, O. C. & Vreugdenhil, E. Glucocorticoid Transcription factor interactions in genomic nuclear
4950–4960 (2014). signaling and stress-​related limbic susceptibility receptor function. Epigenomics 3, 471–485 (2011).

Nature Reviews | Endocrinology


Reviews

89. So, A. Y., Chaivorapol, C., Bolton, E. C., Li, H. 109. Spiegel, K., Leproult, R. & Van Cauter, E. Impact of 131. Johannsson, G. et al. Adrenal insufficiency: review
& Yamamoto, K. R. Determinants of cell- and sleep debt on metabolic and endocrine function. of clinical outcomes with current glucocorticoid
gene-specific transcriptional regulation by the Lancet 354, 1435–1439 (1999). replacement therapy. Clin. Endocrinol. 82, 2–11
glucocorticoid receptor. PLOS Genet. 3, e94 (2007). 110. Hauner, H., Schmid, P. & Pfeiffer, E. F. Glucocorticoids (2015).
90. Zalachoras, I., Houtman, R. & Meijer, O. C. and insulin promote the differentiation of human 132. Lovas, K., Loge, J. H. & Husebye, E. S. Subjective
Understanding stress-​effects in the brain via adipocyte precursor cells into fat cells. J. Clin. health status in Norwegian patients with Addison’s
transcriptional signal transduction pathways. Endocrinol. Metab. 64, 832–835 (1987). disease. Clin. Endocrinol. 56, 581–588 (2002).
Neuroscience 242, 97–109 (2013). 111. Dallman, M. F. et al. Glucocorticoids, chronic stress, 133. Feek, C. M. et al. Patterns of plasma cortisol and
91. Sarabdjitsingh, R. A. et al. Stress responsiveness and obesity. Prog. Brain Res. 153, 75–105 (2006). ACTH concentrations in patients with Addison’s
varies over the ultradian glucocorticoid cycle in a 112. Tsigos, C. et al. Dose-​dependent effects of recombinant disease treated with conventional corticosteroid
brain-​region-specific manner. Endocrinology 151, human interleukin-6 on glucose regulation. J. Clin. replacement. Clin. Endocrinol. 14, 451–458 (1981).
5369–5379 (2010). Endocrinol. Metab. 82, 4167–4170 (1997). 134. Isidori, A. M. et al. Effect of once-​daily, modified-​
92. Sarabdjitsingh, R. A. et al. Ultradian corticosterone 113. McEwen, B. S. Sleep deprivation as a neurobiologic release hydrocortisone versus standard glucocorticoid
pulses balance glutaminergic transmission and and physiologic stressor: allostasis and allostatic load. therapy on metabolism and innate immunity in
synaptic plasticity. Proc. Natl Acad. Sci. USA 111, Metabolism 55, S20–S23 (2006). patients with adrenal insufficiency (DREAM): a single-​
14265–14270 (2014). 114. Zhu, B., Shi, C., Park, C. G., Zhao, X. & Reutrakul, S. blind, randomised controlled trial. Lancet Diabetes
93. Kalafatakis, K. et al. Ultradian rhythmicity of plasma Effects of sleep restriction on metabolism-​related Endocrinol. 6, 173–185 (2018).
cortisol is necessary for normal emotional and parameters in healthy adults: a comprehensive review 135. Bancos, I. et al. Primary adrenal insufficiency is
cognitive responses in man. Proc. Natl Acad. Sci. USA and meta-​analysis of randomized controlled trials. associated with impaired natural killer cell function:
115, E4091–E4100 (2018). Sleep Med. Rev. 45, 18–30 (2019). a potential link to increased mortality. Eur. J.
94. Gjerstad, J. K., Lightman, S. L. & Spiga, F. Role of 115. Gavrila, A. et al. Diurnal and ultradian dynamics of Endocrinol. 176, 471–480 (2017).
glucocorticoid negative feedback in the regulation serum adiponectin in healthy men: comparison with 136. Bjanesoy, T. E. et al. Altered DNA methylation profile
of HPA axis pulsatility. Stress 21, 403–416 (2018). leptin, circulating soluble leptin receptor, and cortisol in Norwegian patients with autoimmune Addison’s
95. Bornstein, S. R., Engeland, W. C., Ehrhart-​Bornstein, M. patterns. J. Clin. Endocrinol. Metab. 88, 2838–2843 disease. Mol. Immunol. 59, 208–216 (2014).
& Herman, J. P. Dissociation of ACTH and (2003). 137. Langenheim, J., Ventz, M., Hinz, A. & Quinkler, M.
glucocorticoids. Trends Endocrinol. Metab. 19, 116. Knutson, K. L. & Van Cauter, E. Associations between Modified-​release prednisone decreases complaints
175–180 (2008). sleep loss and increased risk of obesity and diabetes. and fatigue compared to standard prednisolone in
96. Silverman, M. N., Miller, A. H., Biron, C. A. Ann. NY Acad. Sci. 1129, 287–304 (2008). patients with adrenal insufficiency. Horm. Metab. Res.
& Pearce, B. D. Characterization of an interleukin-6- 117. Adam, T. C. & Epel, E. S. Stress, eating and the reward 45, 96–101 (2013).
and adrenocorticotropin-​dependent, immune-to- system. Physiol. Behav. 91, 449–458 (2007). 138. Mallappa, A. et al. A phase 2 study of Chronocort, a
adrenal pathway during viral infection. Endocrinology 118. Young, E. A., Carlson, N. E. & Brown, M. B. Twenty-​ modified-​release formulation of hydrocortisone, in the
145, 3580–3589 (2004). four-hour ACTH and cortisol pulsatility in depressed treatment of adults with classic congenital adrenal
97. Franchimont, D. et al. Adrenal cortical activation in women. Neuropsychopharmacology 25, 267–276 hyperplasia. J. Clin. Endocrinol. Metab. 100,
murine colitis. Gastroenterology 119, 1560–1568 (2001). 1137–1145 (2015).
(2000). 119. Heuser, I., Yassouridis, A. & Holsboer, F. The combined 139. Lovas, K. & Husebye, E. S. Continuous subcutaneous
98. Viblanc, V. A. et al. An integrative appraisal of the dexamethasone/CRH test: a refined laboratory test for hydrocortisone infusion in Addison’s disease. Eur. J.
hormonal and metabolic changes induced by acute psychiatric disorders. J. Psychiatr. Res. 28, 341–356 Endocrinol. 157, 109–112 (2007).
stress using king penguins as a model. Gen. Comp. (1994). 140. Venneri, M. A. et al. Circadian rhythm of glucocorticoid
Endocrinol. 269, 1–10 (2018). 120. Ising, M. et al. The combined dexamethasone/CRH administration entrains clock genes in immune cells:
99. Cruz-​Topete, D. & Cidlowski, J. A. One hormone, test as a potential surrogate marker in depression. a DREAM trial ancillary study. J. Clin. Endocrinol.
two actions: anti- and pro-​inflammatory effects of Prog. Neuropsychopharmacol. Biol. Psychiatry 29, Metab. 103, 2998–3009 (2018).
glucocorticoids. Neuroimmunomodulation 22, 20–32 1085–1093 (2005). 141. Oksnes, M. et al. Continuous subcutaneous
(2015). 121. Krishnan, V. & Nestler, E. J. The molecular neurobiology hydrocortisone infusion versus oral hydrocortisone
100. Biddie, S. C., Conway-​Campbell, B. L. & Lightman, S. L. of depression. Nature 455, 894–902 (2008). replacement for treatment of Addison’s disease: a
Dynamic regulation of glucocorticoid signalling in 122. Miller, A. H. Depression and immunity: a role for randomized clinical trial. J. Clin. Endocrinol. Metab.
health and disease. Rheumatology 51, 403–412 T cells? Brain Behav. Immun. 24, 1–8 (2010). 99, 1665–1674 (2014).
(2012). 123. Koo, J. W. & Duman, R. S. IL-1beta is an essential 142. Riedel, M., Wiese, A., Schurmeyer, T. H. & Brabant, G.
101. Miller, G. E., Cohen, S. & Ritchey, A. K. Chronic mediator of the antineurogenic and anhedonic effects of Quality of life in patients with Addison’s disease:
psychological stress and the regulation of pro-​ stress. Proc. Natl Acad. Sci. USA 105, 751–756 (2008). effects of different cortisol replacement modes.
inflammatory cytokines: a glucocorticoid-​resistance 124. Horowitz, M. A., Zunszain, P. A., Anacker, C., Exp. Clin. Endocrinol. 101, 106–111 (1993).
model. Health Psychol. 21, 531–541 (2002). Musaelyan, K. & Pariante, C. M. Glucocorticoids and 143. van Staa, T. P. et al. Use of oral corticosteroids in the
102. Oster, H. et al. The functional and clinical significance inflammation: a double-​headed sword in depression? United Kingdom. QJM 93, 105–111 (2000).
of the 24-hour rhythm of circulating glucocorticoids. How do neuroendocrine and inflammatory pathways 144. Overman, R. A., Yeh, J. Y. & Deal, C. L. Prevalence
Endocr. Rev. 38, 3–45 (2017). interact during stress to contribute to the of oral glucocorticoid usage in the United States:
103. Keller, M. et al. A circadian clock in macrophages pathogenesis of depression? Mod. Trends a general population perspective. Arthritis Care Res.
controls inflammatory immune responses. Proc. Natl Pharmacopsychiatry 28, 127–143 (2013). 65, 294–298 (2013).
Acad. Sci. USA 106, 21407–21412 (2009). 125. Munhoz, C. D. et al. Chronic unpredictable stress 145. Curtis, J. R. et al. Population-​based assessment
104. Boivin, D. B. et al. Circadian clock genes oscillate in exacerbates lipopolysaccharide-​induced activation of adverse events associated with long-​term
human peripheral blood mononuclear cells. Blood of nuclear factor-​kappaB in the frontal cortex and glucocorticoid use. Arthritis Rheum. 55, 420–426
102, 4143–4145 (2003). hippocampus via glucocorticoid secretion. J. Neurosci. (2006).
105. Koo, J. W., Russo, S. J., Ferguson, D., Nestler, E. J. 26, 3813–3820 (2006). 146. McDonough, A. K., Curtis, J. R. & Saag, K. G.
& Duman, R. S. Nuclear factor-​kappaB is a critical 126. Pariante, C. M. Glucocorticoid receptor function The epidemiology of glucocorticoid-​associated
mediator of stress-​impaired neurogenesis and in vitro in patients with major depression. Stress 7, adverse events. Curr. Opin. Rheumatol. 20, 131–137
depressive behavior. Proc. Natl Acad. Sci. USA 107, 209–219 (2004). (2008).
2669–2674 (2010). 127. Kenis, G. & Maes, M. Effects of antidepressants on the 147. Leung, D. Y. & Bloom, J. W. Update on glucocorticoid
106. Pace, T. W., Hu, F. & Miller, A. H. Cytokine-​effects production of cytokines. Int. J. Neuropsychopharmacol. action and resistance. J. Allergy Clin. Immunol. 111,
on glucocorticoid receptor function: relevance to 5, 401–412 (2002). 3–22 (2003).
glucocorticoid resistance and the pathophysiology and 128. Bjornsdottir, S. et al. Drug prescription patterns in
treatment of major depression. Brain Behav. Immun. patients with Addison’s disease: a Swedish population-​ Author contributions
21, 9–19 (2007). based cohort study. J. Clin. Endocrinol. Metab. 98, The authors contributed equally to all aspects of the article.
107. Pace, T. W. et al. Increased stress-​induced 2009–2018 (2013).
inflammatory responses in male patients with major 129. Dunlop, D. Eighty-​six cases of Addison’s disease. BMJ Competing interests
depression and increased early life stress. Am. J. 2, 887–891 (1963). The authors declare no competing interests.
Psychiatry 163, 1630–1633 (2006). 130. Giordano, R. et al. Metabolic and cardiovascular profile
108. Cohen, S. et al. Chronic stress, glucocorticoid receptor in patients with Addison’s disease under conventional Publisher’s note
resistance, inflammation, and disease risk. Proc. Natl glucocorticoid replacement. J. Endocrinol. Invest. 32, Springer Nature remains neutral with regard to jurisdictional
Acad. Sci. USA 109, 5995–5999 (2012). 917–923 (2009). claims in published maps and institutional affiliations.

www.nature.com/nrendo

You might also like